Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Fungi (Basel) ; 6(3)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971819

ABSTRACT

Cutaneous invasive fungal wound infections after life-threatening dismounted complex blast injury (DCBI) and natural disasters complicate clinical care. These wounds often require aggressive repeated surgical debridement, can result in amputations and hemipelvectomies and have a 38% mortality rate. Given the substantial morbidity associated with cutaneous fungal wound infections, patients at risk need immediate empiric treatment mandating the use of rapidly acting broad-spectrum antimicrobials, acting on both fungi and bacteria, that are also effective against biofilm and can be administered topically. Designed antimicrobial peptides (dAMPs) are engineered analogues of innate antimicrobial peptides which provide the first line of defense against invading pathogens. The antifungal and antibacterial effect and mammalian cytotoxicity of seven innovative dAMPs, created by iterative structural analog revisions and physicochemical and functional testing were investigated. The dAMPs possess broad-spectrum antifungal activity, in addition to being effective against Gram-negative and Gram-positive bacteria, which is crucial as many wounds are polymicrobial and require immediate empiric treatment. Three of the most potent dAMPs-RP504, RP556 and RP557-possess limited mammalian cytotoxicity following 8 h incubation. If these encouraging broad-spectrum antimicrobial and rapid acting results are translated clinically, these novel dAMPs may become a first line empiric topical treatment for traumatic wound injuries.

2.
Antibiotics (Basel) ; 9(1)2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31940992

ABSTRACT

Acne vulgaris, caused by the Gram-positive bacterium Cutibacterium acnes, is a prevalent dermatologic condition with substantial cutaneous and psychological morbidity. Mild acne is treated with topical antibiotics with more severe inflammatory forms requiring the prolonged use of oral antibiotics, resulting in antimicrobial resistance development. Innovative treatment alternatives, providing complete microbicidal eradication with minimal safety issues and limited susceptibility to microbial resistance, are fervently sought. Designed antimicrobial peptides (dAMPs) are engineered analogs of naturally occurring AMPs that possess a reduced likelihood of developing bacterial resistance. Seven novel dAMP sequences were screened for in vitro bactericidal effectiveness against antibiotic resistant C. acnes clinical isolates. Five peptides (RP444, RP551, RP554, RP556, and RP557) exhibited potent in vitro antibacterial activity. The Therapeutic Index, a measure of specificity for killing multidrug resistant C. acnes over mammalian cells, was determined using bioluminescent human keratinocytes. The Therapeutic Index was highest for the disulfide dAMP, RP556, with a value of 130. The lead dAMP candidate RP556, was further evaluated in a multidrug-resistant C. acnes intradermal murine infection model. A topical application of 5 mg/mL RP556 (0.5%) eliminated infection. If these preclinical results are translated clinically, dAMPs may become a viable topical monotherapy for the treatment of recalcitrant acne infections.

3.
Article in English | MEDLINE | ID: mdl-31451496

ABSTRACT

Recurrent vulvovaginal candidiasis (RVVC) is a widespread chronic infection that has a substantial negative impact on work and quality of life. The development of antimicrobial resistance and biofilm formation are speculated to contribute to Candida pathogenicity and treatment ineffectiveness. Designed antimicrobial peptides (dAMPs) are chemically modified from endogenous antimicrobial peptides that provide the first line of defense against pathogens. The goal here is to identify a dAMP for the topical treatment of RVVC. The dAMP MICs were determined for 46 fluconazole-susceptible and fluconazole-resistant Candida spp. clinical isolates. The possibility of inducing dAMP drug resistance and comparison of dAMP and fluconazole activity against preformed Candida biofilm and biofilm formation were evaluated. Assessment of mammalian cell viability was determined using bioluminescent human keratinocytes. The dAMP effect on fungus was probed via scanning electron microscopy, and topically applied dAMP activity was evaluated in a rodent vulvovaginal candidiasis (VVC) infection model. dAMPs demonstrated broad-spectrum antimicrobial activity against common causative clinical Candida isolates, reduced preformed biofilm, and inhibited biofilm formation. An evaluated dAMP did not induce resistance after repeated exposure of Candida tropicalis The dAMPs were selective for Candida cells with limited mammalian cytotoxicity with substantial activity in a rodent VVC model. dAMPs are described as having potent antifungal and antibiofilm activity, likely direct membrane action with selectivity for Candida cells, with limited resistance development. Combined with activity in a rodent VVC model, the data support clinical evaluation of dAMPs for topical treatment of VCC and recurrent VVC infections.


Subject(s)
Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Candida/drug effects , Candidiasis, Vulvovaginal/drug therapy , Peptides/pharmacology , Animals , Biofilms/drug effects , Cell Survival/drug effects , Drug Resistance, Fungal/drug effects , Female , Fluconazole/pharmacology , Humans , Keratinocytes/microbiology , Microbial Sensitivity Tests/methods , Rats , Rats, Wistar
4.
Invest Ophthalmol Vis Sci ; 58(14): 6273-6281, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29242901

ABSTRACT

Purpose: To limit corneal damage and potential loss of vision, bacterial keratitis must be treated aggressively. Innovation in antimicrobials is required due to the need for empirical treatment and the rapid emergence of bacterial resistance. Designed host defense peptides (dHDPs) are synthetic analogues of naturally occurring HDPs, which provide defense against invading pathogens. This study investigates the use of novel dHDPs for the treatment of bacterial keratitis. Methods: The minimum inhibitory concentrations (MICs) were determined for dHDPs on both Gram-positive and -negative bacteria. The minimum biofilm eradication concentrations (MBEC) and in vitro time-kill assays were determined. The most active dHDP, RP444, was evaluated for propensity to induce drug resistance and therapeutic benefit in a murine Pseudomonas aeruginosa keratitis model. Results: Designed HDPs were bactericidal with MICs ranging from 2 to >64 µg/mL and MBEC ranging from 6 to 750 µg/mL. In time-kill assays, dHDPs were able to rapidly reduce bacterial counts upon contact with as little as 2 µg/mL. RP444 did not induce resistance after repeated exposure of P. aeruginosa to subinhibitory concentrations. RP444 demonstrated significant efficacy in a murine model of bacterial keratitis as evidenced by a significant dose-dependent decrease in ocular clinical scores, a significantly reduced bacterial load, and substantially decreased inflammatory cell infiltrates. Conclusions: Innovative dHDPs demonstrated potent antimicrobial activity, possess a limited potential for development of resistance, and reduced the severity of murine P. aeruginosa keratitis. These studies demonstrate that a novel dHDP may have potential to treat patients with sight-threatening bacterial keratitis.


Subject(s)
Biofilms/drug effects , Cornea/microbiology , Eye Infections, Bacterial/drug therapy , Keratitis/drug therapy , Organotechnetium Compounds/administration & dosage , Peptides, Cyclic/administration & dosage , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/isolation & purification , Animals , Culture Media, Serum-Free , Disease Models, Animal , Dose-Response Relationship, Drug , Eye Infections, Bacterial/microbiology , Keratitis/microbiology , Mice, Inbred C57BL , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects
5.
Pharmacol Res ; 113(Pt A): 62-70, 2016 11.
Article in English | MEDLINE | ID: mdl-27521836

ABSTRACT

Erythropoietin (EPO) has been shown to protect the heart against acute myocardial infarction in pre-clinical studies, however, EPO failed to reduce infarct size in clinical trials and showed significant safety problems. Here, we investigated cardioprotective effects of two selective non-erythropoietic EPO receptor ligand dimeric peptides (AF41676 and AF43136) lacking erythropoietic activity, EPO, and the prolonged half-life EPO analogue, darbepoetin in acute myocardial infarction (AMI) in rats. In a pilot study, EPO at 100U/mL significantly decreased cell death compared to vehicle (33.8±2.3% vs. 40.3±1.5%, p<0.05) in rat neonatal cardiomyocytes subjected to simulated ischemia/reperfusion. In further studies (studies 1-4), in vivo AMI was induced by 30min coronary occlusion and 120min reperfusion in male Wistar rats. Test compounds and positive controls for model validation (B-type natriuretic peptide, BNP or cyclosporine A, CsA) were administered iv. before the onset of reperfusion. Infarct size (IS) was measured by standard TTC staining. In study 1, 5000U/kg EPO reduced infarct size significantly compared to vehicle (45.3±4.8% vs. 59.8±4.5%, p<0.05). In study 2, darbepoetin showed a U-shaped dose-response curve with maximal infarct size-reducing effect at 5µg/kg compared to the vehicle (44.4±5.7% vs. 65.9±2.7%, p<0.01). In study 3, AF41676 showed a U-shaped dose-response curve, where 3mg/kg was the most effective dose compared to the vehicle (24.1±3.9% vs. 44.3±2.5%, p<0.001). The positive control BNP significantly decreased infarct size in studies 1-3 by approximately 35%. In study 4, AF43136 at 10mg/kg decreased infarct size, similarly to the positive control CsA compared to the appropriate vehicle (39.4±5.9% vs. 58.1±5.4% and 45.9±2.4% vs. 63.8±4.1%, p<0.05, respectively). This is the first demonstration that selective, non-erythropoietic EPO receptor ligand dimeric peptides AF41676 and AF43136 administered before reperfusion are able to reduce infarct size in a rat model of AMI. Therefore, non-erythropoietic EPO receptor peptide ligands may be promising cardioprotective agents.


Subject(s)
Cardiotonic Agents/pharmacology , Erythropoietin/metabolism , Myocardial Infarction/drug therapy , Myocardium/metabolism , Animals , Ligands , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/pharmacology , Pilot Projects , Rats , Rats, Wistar
6.
Drug Metab Dispos ; 41(4): 774-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23318685

ABSTRACT

Peginesatide, a polyethylene glycol (PEG)ylated peptide-based erythropoiesis-stimulating agent, stimulates the erythropoietin receptor dimer that governs erythropoiesis. Studies were designed to determine the erythropoietic response, pharmacokinetics (PK), tissue distribution, metabolism, and excretion of peginesatide in nonhuman primates following a single i.v. dose. The PK profile of peginesatide (0.1-5 mg/kg) is characterized by low, dose-dependent plasma clearance; small volume of distribution; and long half-life. The peginesatide PK profile following a single i.v. dose is consistent with the sustained erythropoiesis. Biodistribution quantitative whole-body autoradiography demonstrated high peginesatide levels in bone marrow (i.e., primary hematopoietic site) as well as other known hematopoietic sites persisting through at least 3 weeks at 2.1 mg/kg. Microautoradiography analysis at 48 hours postdose revealed uniform and high distribution of radioactivity in the bone marrow and splenic red pulp with less extensive distribution in the renal cortex (glomeruli, associated ducts, interstitial cells). Radioactivity in the kidney was most prominent in the outer medullary and papillary interstitium. At 2 weeks after dosing, cumulative radioactivity recovery in the urine and feces was 60 and 7% of the administered dose, respectively, with most of the radioactivity associated with the parent molecule. In conclusion, the PK characteristics are consistent with a PEGylated peptide of a 45-kDa molecular mass, specifically low volume of distribution and long half-life. Drug was localized principally to hematopoietic sites, and nonspecific tissue retention was not observed. The nonhuman primate data indicate that peginesatide is metabolically stable and primarily excreted in the urine.


Subject(s)
Hematinics/administration & dosage , Hematinics/pharmacokinetics , Peptides/administration & dosage , Peptides/pharmacokinetics , Administration, Intravenous , Animals , Biological Availability , Bone Marrow/diagnostic imaging , Bone Marrow/metabolism , Dose-Response Relationship, Drug , Erythropoiesis/drug effects , Hematinics/metabolism , Hematinics/pharmacology , Kidney/diagnostic imaging , Kidney/metabolism , Macaca fascicularis , Male , Peptides/metabolism , Peptides/pharmacology , Radionuclide Imaging , Spleen/diagnostic imaging , Spleen/metabolism
7.
Xenobiotica ; 42(7): 660-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22188389

ABSTRACT

The pharmacokinetics (PK) (absorption, distribution, metabolism, excretion) of peginesatide, a synthetic, PEGylated, investigational, peptide-based erythropoiesis-stimulating agent (ESA), was evaluated in rats. The PK profile was evaluated at 0.1-5 mg·kg(-1) IV using unlabeled or [(14)C]-labeled peginesatide. Mass balance, tissue distribution and metabolism were evaluated following IV administration of 5 mg·kg(-1) [(14)C]-peginesatide, with tissue distribution also evaluated by quantitative whole-body autoradiography (QWBA) following an IV dose of 17 mg·kg(-1) [(14)C]-peginesatide. Plasma clearance was slow and elimination was biphasic with unchanged peginesatide representing >90% of the total radioactivity of the total radioactive exposure. Slow uptake of the radiolabeled compound from the vascular compartment into the tissues was observed. Biodistribution to bone marrow and extramedullary hematopoietic sites, and to highly vascularized lymphatic and excretory tissues occurred. A predominant degradation event to occur in vivo was the loss of one PEG chain from the branched PEG moiety to generate mono-PEG. Renal excretion was the primary mechanism (41%) of elimination, with parent molecule (67%) the major moiety excreted. In conclusion, elimination of [(14)C]-peginesatide-derived radioactivity was extended, retention preferentially occurred at sites of erythropoiesis (bone marrow), and urinary excretion was the primary elimination route.


Subject(s)
Hematinics/pharmacokinetics , Peptides/pharmacokinetics , Absorption/physiology , Animals , Autoradiography , Hematinics/urine , Male , Metabolic Clearance Rate , Peptides/urine , Rats , Rats, Sprague-Dawley , Tissue Distribution
8.
Drug Chem Toxicol ; 34(3): 240-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21649477

ABSTRACT

Peginesatide is a PEGylated, investigational, peptide-based erythropoiesis-stimulating agent (ESA) that was designed and engineered to stimulate specifically the erythropoietin receptor dimer that governs erythropoiesis. Clinical use of peginesatide is anticipated to result in chronic dosing in chronic kidney disease (CKD) patients, and the nonclinical data to support development should include an evaluation of carcinogenic potential evaluation. Peginesatide was not mutagenic or clastogenic in a standard genotoxicity battery of tests. Doses for a rasH2 transgenic mouse carcinogenicity assay were defined in a 28-day study in the wild-type littermates of the rasH2 transgenic mouse strain, using intravenous doses of 1-25 mg/kg on days 1 and 22. The findings were consistent with exaggerated pharmacology, including polycythemia, with associated increases in hemoglobin level and extramedullary hematopoiesis and bone marrow hypercellularity.


Subject(s)
Carcinogens/toxicity , Mutagens/toxicity , Peptides/toxicity , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , CHO Cells , Carcinogenicity Tests , Carcinogens/pharmacokinetics , Chromosome Aberrations/chemically induced , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Escherichia coli/genetics , Female , Genes, ras , Humans , Injections, Intravenous , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Mutagenicity Tests , Mutagens/pharmacokinetics , Peptides/pharmacokinetics , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Toxicity Tests, Chronic
9.
Birth Defects Res B Dev Reprod Toxicol ; 89(2): 155-63, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437475

ABSTRACT

BACKGROUND: Aperi- and postnatal reproduction toxicity study was conducted in rats treated with Hematide, a synthetic PEGylated peptidic erythropoiesis stimulating agent (ESA). METHODS: Hematide, at IV doses of 0, 0.5, 3, and 15 mg/kg, was administered from implantation through lactation on gestation days (GDs) 5 and 18 and lactation day (LD) 13. RESULTS: Hematide induced pronounced polycythemia in all Hematide-treated dams. On LDs 2 and 21, hemoglobin (Hgb) increases above control levels were 3.1, 5.2, and 5.0 g/dL and 4.1, 5.1, and 5.5 g/dL at the 0.5, 3, and 15 mg/kg/dose, respectively. There were no effects on parturition, lactation, or maternal behavior in the F0 generation female rats. A slight decrease in pup viability on postpartum days 2-4 and lower body weights and/or body weight gain for the F1 generation were associated with pronounced polycythemia and decreases in maternal body weight gain and/or food consumption at > or =3 mg/kg/dose. Hematide fetal exposure was negligible. No Hematide effect, other than on growth and survival, was noted on developmental, functional, mating, and fertility end points in the F1 generation rats, and no effect on litter or fetal parameters was observed in the F2 generation. The maternal no-observed-adverse-effect level (NOAEL) for Hematide was 0.5 mg/kg, and the NOAEL for parturition and maternal behavior was 15 mg/kg. The NOAEL for F1 pup viability and growth was 0.5 mg/kg/dose. CONCLUSIONS: In conclusion, the Hematide-associated adverse findings were attributed to exaggerated erythropoiesis (pronounced and prolonged polycythemia) resulting from administration of an ESA to pregnant animals.


Subject(s)
Hematinics/toxicity , Maternal Exposure , Peptides/toxicity , Polyethylene Glycols/toxicity , Reproduction/drug effects , Teratogens/toxicity , Animals , Animals, Newborn/growth & development , Behavior, Animal/drug effects , Body Weight/drug effects , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Hematinics/classification , Injections, Intravenous , Lactation/drug effects , Longevity/drug effects , Male , Maternal Behavior/drug effects , Peptides/classification , Polycythemia/chemically induced , Polycythemia/physiopathology , Polyethylene Glycols/classification , Pregnancy , Rats , Reproduction/physiology , Teratogens/classification
10.
Drug Chem Toxicol ; 33(1): 28-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19995307

ABSTRACT

The subchronic toxicity of Hematide™, a synthetic PEGylated peptidic erythropoiesis-stimulating agent (ESA), was evaluated in CD-1 mice at intravenous doses of 0, 1, 5, 25, and 125 mg/kg administered once every 3 weeks for 3 months. Hematide displayed sustained plasma levels with reduced clearance and prolonged half-lives up to 59.4 hours that translated into sustained, pronounced polycythemia, bone marrow hyperplasia, and splenic and liver extramedullary hematopoiesis. Toxicological findings were considered to be secondary to exaggerated pharmacology, rather than a direct drug effect, and included mortality at ≥25 mg/kg/dose. The no-observed-adverse-effect-level was determined to be 5 mg/kg.


Subject(s)
Peptides/toxicity , Polyethylene Glycols/toxicity , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Dose-Response Relationship, Drug , Female , Half-Life , Hematopoiesis, Extramedullary/drug effects , Hyperplasia/chemically induced , Male , Mice , No-Observed-Adverse-Effect Level , Peptides/administration & dosage , Peptides/pharmacokinetics , Polycythemia/chemically induced , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics
11.
Basic Clin Pharmacol Toxicol ; 104(2): 155-63, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19143750

ABSTRACT

Hematide is a synthetic peptide-based, PEGylated erythropoiesis-stimulating agent, which is being developed for the chronic treatment of anaemia associated with chronic renal failure. To support the safety of long-term dosing of chronic renal failure patients, a comprehensive toxicology programme was implemented including rat subchronic and chronic studies. Rats were administered 0, 0.1, 1 and 10 mg/kg of Hematide every 3 weeks for 3 months via subcutaneous injection or for 6 months via intravenous injection. The dosing period was followed by a 6-week follow-up period. The primary pharmacology of Hematide resulted in erythroid polycythemia as measured by elevated haemoglobin levels that were time- and dose-dependent. The pharmacology profiles were similar regardless of administration route. For example, for male rats at Day 90, subcutaneous dosing resulted in haemoglobin increases of 2.7, 4.5 and 6.9 g/dl for 0.1, 1 and 10 mg Hematide/kg respectively, compared to 2.8, 5.7 and 7.4 g/dl increases for intravenous dosing. Histopathological changes were related to the prolonged severe polycythemia induced in normocythemic animals administered an erythropoiesis-stimulating agent. The findings included extramedullary haematopoiesis in the spleen and liver, bone marrow hypercellularity and organ congestion. Microscopic findings were reversible, demonstrating a return towards control findings within 6 weeks following cessation of dosing. Systemic exposures, based on both area under the curve (AUC) and maximum concentration (C(max)), were substantially greater for intravenous than subcutaneous administration. No Hematide-specific antibodies were detected. In conclusion, Hematide is a potent erythropoiesis-stimulating agent, and the studies provide support for the safety of clinical development, including chronic dosing, for the treatment of anaemia associated with chronic renal failure.


Subject(s)
Hematinics/adverse effects , Hematinics/pharmacology , Peptides/adverse effects , Peptides/pharmacology , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematinics/administration & dosage , Hematinics/pharmacokinetics , Hematopoiesis, Extramedullary/drug effects , Hemoglobins/analysis , Injections, Intravenous , Injections, Subcutaneous , Male , Organ Size/drug effects , Peptides/administration & dosage , Peptides/pharmacokinetics , Polycythemia/chemically induced , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Rats , Rats, Sprague-Dawley
12.
Haematologica ; 93(9): 1376-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18519511

ABSTRACT

Hematide is a synthetic peptide-based, pegylated erythropoiesis stimulating agent in clinical development for treatment of anemia. To support chronic clinical dosing requirements, a 9-month repeat dose IV monkey safety study was undertaken. Animals received 0, 0.2, 2 or 20 mg/kg hematide IV every three weeks for nine months followed by a 14-week recovery. Hematide administration was associated with time and dose-dependent polycythemia. Histological findings were related to exaggerated pharmacology that was secondary to the administration of an erythropoiesis stimulating agent to a normocythemic animal. In conclusion, these results support the use of repeated administration of hematide for the correction of anemia.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Hematinics/pharmacology , Macaca fascicularis , Peptides/pharmacology , Polyethylene Glycols/pharmacology , Animals , Disease , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Erythropoiesis/drug effects , Peptides/adverse effects , Polyethylene Glycols/adverse effects
13.
Drug Chem Toxicol ; 31(2): 229-44, 2008.
Article in English | MEDLINE | ID: mdl-18330784

ABSTRACT

The pharmacology, toxicokinetics, and safety of Hematide, a synthetic peptidic erythropoiesis-stimulating agent (ESA), were characterized. Hematide was given intravenously (0, 0.5, 5, and 50 mg/kg) weekly for five weeks with a 6- (rat) and 12-week (monkey) recovery period. The pharmacological action of Hematide resulted in polycythemia. Histopathology consistent with drug-induced exaggerated pharmacology was observed primarily in rats. Secondary sequelae resulting from pronounced polycythemia was considered the cause of deaths in rats and a single high-dose monkey. Toxicokinetic analysis indicated prolonged exposure. In conclusion, Hematide is a potent ESA and the safety and efficacy profile support clinical development.


Subject(s)
Erythropoiesis/drug effects , Peptides/administration & dosage , Peptides/toxicity , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/toxicity , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Injections, Intravenous , Macaca fascicularis , Male , Peptides/pharmacokinetics , Polycythemia/chemically induced , Polyethylene Glycols/pharmacokinetics , Rats , Rats, Sprague-Dawley , Species Specificity
14.
Exp Hematol ; 35(8): 1201-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17662888

ABSTRACT

OBJECTIVE: To evaluate the potential of Hematide, a PEGylated, synthetic peptide-based erythropoiesis-stimulating agent that is in clinical development for the treatment of anemia associated with chronic kidney disease and cancer, to correct antierythropoietin antibody-associated pure red cell aplasia (PRCA). MATERIALS AND METHODS: The binding of anti-Hematide antibodies (mouse, rabbit, and monkey) to recombinant human erythropoietin (rHuEPO) and of anti-rHuEPO antibodies (mouse, goat, rat, and human) to Hematide were evaluated. An anti-EPO antibody-mediated anemia rat model was developed by subcutaneously administering rHuEPO to rats three times weekly for 4 weeks. Sixty percent of the animals developed PRCA as characterized by severe anemia, reduced reticulocytes, anti-EPO antibodies, and limited bone marrow erythroid precursors. The effect of Hematide administration on the PRCA rats was evaluated. RESULTS: Antibodies to EPO do not cross react with Hematide and, conversely, antibodies to Hematide do not cross react with EPO. Hematide corrected antibody-induced anemia in a rat PRCA model. CONCLUSIONS: The data support the potential of Hematide to correct anti-EPO antibody-associated PRCA in humans. In addition, the data suggest a negligible risk for development of anti-EPO antibody-induced PRCA secondary to Hematide administration.


Subject(s)
Anemia/drug therapy , Erythropoietin/immunology , Peptides/pharmacology , Polyethylene Glycols/pharmacology , Red-Cell Aplasia, Pure/drug therapy , Anemia/etiology , Animals , Antibodies , Bone Marrow/pathology , Bone Marrow/physiopathology , Colony-Forming Units Assay , Disease Models, Animal , Erythropoietin/pharmacology , Erythropoietin/therapeutic use , Humans , Peptides/immunology , Rabbits , Rats , Red-Cell Aplasia, Pure/pathology
15.
Exp Hematol ; 34(10): 1303-11, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16982323

ABSTRACT

OBJECTIVE: To evaluate the preclinical erythropoiesis stimulating properties of Hematide, a novel, PEGylated, synthetic peptide for the treatment of anemia associated with chronic kidney disease and cancer. METHODS: The in vitro activity of Hematide was assessed in competitive binding, proliferation, signal transduction, and apoptosis assays, and in erythroid colony-forming assays with CD34(+) cells purified from human bone marrow. Erythropoiesis and pharmacokinetics were evaluated in rat, monkey, and a rat chronic renal insufficiency (CRI) model following single administration. Erythropoiesis and immunogenicity were also evaluated following repeat administration in rats. RESULTS: Hematide binds and activates the erythropoietin receptor and causes proliferation and differentiation of erythroid progenitor cells. Sustained circulatory persistence of Hematide is observed in rats and monkeys. In a rat CRI model, Hematide exhibited twofold lower clearance than in the normal rat, with hypothesis consistent with Hematide being cleared, at least partially, via the kidney. A dose-dependent rise in hemoglobin (Hgb) and duration of response was observed following single administration in rats and monkeys. Hematide was able to alleviate anemia in an experimental CRI rodent model. Repeat intravenous (IV) and subcutaneous (SC) administration in rats yielded similar erythrogenic responses, with no anti-Hematide antibodies being detected. CONCLUSIONS: Hematide is a potent erythropoiesis stimulating agent with a prolonged half-life and slow clearance times. It is anticipated that similar prolonged clearance and activity will be observed in the clinic, potentially enabling dosing intervals of 3 to 4 weeks that may translate into improved patient convenience for the treatment of anemia.


Subject(s)
Anemia/drug therapy , Erythropoiesis/drug effects , Peptides/pharmacology , Peptides/pharmacokinetics , Polyethylene Glycols/pharmacology , Polyethylene Glycols/pharmacokinetics , Anemia/etiology , Anemia/immunology , Anemia/metabolism , Animals , Antibodies/immunology , Antibodies/metabolism , Cell Line , Cell Proliferation/drug effects , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Erythroid Precursor Cells/metabolism , Erythropoiesis/immunology , Half-Life , Humans , Kidney/immunology , Kidney/metabolism , Macaca fascicularis , Male , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/immunology , Peptides/immunology , Peptides/therapeutic use , Polyethylene Glycols/therapeutic use , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Erythropoietin/metabolism , Renal Insufficiency/drug therapy , Renal Insufficiency/immunology , Renal Insufficiency/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Stem Cells , Time Factors
16.
Blood ; 108(6): 1830-4, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16720830

ABSTRACT

Hematide is an investigational pegylated synthetic peptide that stimulates erythropoiesis in animal models and is being developed for the treatment of anemia associated with chronic renal failure and cancer. This study evaluated the safety and pharmacodynamics of single, intravenous doses (0.025, 0.05, and 0.1 mg/kg) of Hematide in 28 healthy male volunteers. All doses of Hematide were well tolerated, with safety profiles similar to those of placebo. Hematide showed a dose-dependent increase in reticulocytes. The 0.1-mg/kg dose was associated with a statistically significant increase in hemoglobin (Hgb) from baseline compared to the placebo group (13.6 +/- 3.9 g/L [1.36 +/- 0.39 g/dL] versus 3.9 +/- 3.8 g/L [0.39 +/- 0.38 g/dL]; P < .001) that was sustained for longer than 1 month. These results support phase 2 studies in patients with anemia associated with chronic kidney disease or cancer and suggest that Hematide administered as infrequently as once a month may result in a sustained elevation of Hgb levels. (Please note that Hematide is a proposed trade name; the compound does not yet have a nonproprietary name.).


Subject(s)
Erythropoiesis/drug effects , Peptides/pharmacology , Adult , Anemia/drug therapy , Antibody Formation , Drug Tolerance , Hemoglobins/metabolism , Humans , Male , Peptides/immunology , Peptides/toxicity , Reticulocytes/drug effects , Safety
17.
Catheter Cardiovasc Interv ; 57(3): 387-94, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12410519

ABSTRACT

Photodynamic therapy (PDT) has been approved as a tissue-specific light-activated cytotoxic therapy for many diseases. The ability of PDT to destroy target tissues selectively is especially appealing for atherosclerotic plaque. Biotechnology has developed a new generation of selective photosensitizers and catheter-based technological advances in light delivery have allowed the introduction of PDT into the vasculature. The largest experience to date is with motexafin lutetium (MLu, Antrin), an expanded porphyrin (texaphyrin) that accumulates in plaque. The combination of the motexafin lutetium and endovascular illumination, or Antrin phototherapy, has been shown to reduce plaque in animal models. Antrin phototherapy generates cytotoxic singlet oxygen that has been shown to induce apoptosis in macrophages and smooth muscle cells. The safety, tolerability, and preliminary efficacy of Antrin phototherapy has been assessed in a phase 1 dose-ranging clinical trial in subjects with peripheral artery disease and is currently being examined in a phase 1 study in subjects with lesions of the native coronary arteries undergoing stent implantation. The preliminary results suggest that Antrin phototherapy is safe, well tolerated, and nontraumatic.


Subject(s)
Photochemotherapy , Animals , Coronary Artery Disease/drug therapy , Graft Occlusion, Vascular/drug therapy , Humans , Metalloporphyrins/therapeutic use , Photosensitizing Agents/therapeutic use , Treatment Outcome
18.
Adv Drug Deliv Rev ; 54(7): 1041-8, 2002 Oct 16.
Article in English | MEDLINE | ID: mdl-12384320

ABSTRACT

Poly(ortho esters), POE, are synthetic bioerodible polymers that can be prepared as solid materials, or as viscous, injectable polymers. These materials have evolved through a number of families, and the latest member of this family, POE IV, is particularly well suited to drug delivery since latent acid is integrated into the polymer backbone, thereby, modulating surface erosion. POE IV predominantly undergoes surface erosion and is able to moderate drug release over periods from days to many months. One indication in which the POE IV polymer is currently being investigated is in sustained post-surgical pain management. The local anesthetic agent, mepivacaine, has been incorporated into a viscous, injectable POE IV and its potential to provide longer-acting anesthesia has been explored in non-clinical models.


Subject(s)
Drug Delivery Systems/methods , Pain, Postoperative/drug therapy , Polymers/administration & dosage , Animals , Humans , Polymers/chemistry
19.
Retina ; 22(4): 391-405; quiz 527-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12172104

ABSTRACT

PURPOSE: To review the biophysical basis and current state of therapy for photodynamic closure of subfoveal choroidal neovascularization in the eye. METHODS: A review of the literature is included, which encompasses the chemical structure, biophysical mechanism of action, range of available agents, status of clinical trials, clinical indications, results of treatments, complications, and future directions. RESULTS: Photodynamic therapy has been shown to be effective in closing both experimental choroidal neovascularization in animal models as well as subfoveal choroidal neovascularization in humans. The therapy results in temporary closure of choroidal new vessels for a period of approximately 1 to 4 weeks. By 12 weeks, most patients have reperfusion or reproliferation of choroidal new vessels resulting in the need for retreatment to achieve continued closure and visual stabilization. Differences exist in the quantum yield, clinical efficiency, and light and sensitizer dose requirements between different classes of agents. Further clinical trials will be required to determine the optimal form of therapy, with verteporfin (Visudyne) as the only currently approved agent. Other agents, including tin etiopurpurin (Purlytin) and motexafin lutetium (Optrin), are currently undergoing phase III, and phase II trials, respectively. CONCLUSIONS: Photodynamic therapy is a promising treatment modality shown to be effective in achieving closure and stabilization of vision loss compared with placebo control in eyes with subfoveal choroidal neovascularization.


Subject(s)
Choroidal Neovascularization/drug therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Choroidal Neovascularization/etiology , Clinical Trials as Topic , Humans , Macular Degeneration/complications , Myopia/complications , Photosensitizing Agents/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...